Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Sep 1.
Published in final edited form as: Ophthalmol Retina. 2018 Mar 8;2(9):949–962. doi: 10.1016/j.oret.2018.01.016

The genetics of retinopathy of prematurity: a model for neovascular retinal disease

Ryan Swan 1, Sang Jin Kim 2,3, J Peter Campbell 2, R V Paul Chan 4,5, Kemal Sonmez 6, Kent D Taylor 7, Xiaohui Li 7, Yii-Der Ida Chen 7, Jerome I Rotter 7, Charles Simmons 8, Michael F Chiang 1,2; the Imaging and Informatics in ROP Research Consortium
PMCID: PMC6150458  NIHMSID: NIHMS949815  PMID: 30250936

Abstract

TOPIC

Retinopathy of prematurity (ROP) is a proliferative retinal vascular disease in premature infants, and is a major cause of childhood blindness worldwide. In addition to known clinical risk factors such as low birth weight and gestational age, there is a growing body of evidence supporting a genetic basis for ROP.

CLINICAL RELEVANCE

While comorbidities and environmental factors have been identified as contributing to ROP outcomes in premature infants, most notably gestational age and oxygen, some infants progress to severe disease despite absence of these clinical risk factors. The contribution of genetic factors may explain these differences and allow better detection and treatment of infants at risk for severe ROP.

METHODS

To comprehensively review genetic factors that potentially contribute to the development and severity of ROP, we conducted a literature search focusing on the genetic basis for ROP. Terms related to other heritable retinal vascular diseases like “familial exudative vitreoretinopathy”, as well as to genes implicated in animal models of ROP, were also used to capture research in diseases with similar pathogenesis to ROP in humans with known genetic components.

RESULTS

Contributions across several genetic domains are described including vascular endothelial growth factor, the Wnt signaling pathway, insulin-like growth factor 1, inflammatory mediators, and brain-derived neurotrophic factor.

CONCLUSIONS

Most candidate gene studies of ROP have limitations such as inability to replicate results, conflicting results from various studies, small sample size, and differences in clinical characterization. Additional difficulty arises in separating the contribution of genetic factors like Wnt signaling to ROP and prematurity. Although studies have implicated involvement of multiple signaling pathways in ROP, the genetics of ROP have not been clearly elucidated. Next-generation sequencing and genome-wide association studies have potential to expand future understanding of underlying genetic risk factors and pathophysiology of ROP.

INTRODUCTION

Retinopathy of prematurity (ROP) is a retinal vascular disorder affecting premature low birth weight infants, and is a major cause of childhood blindness in the United States and internationally. Beyond the clinical impact, infancy-acquired visual loss from ROP represents an enormous social and economic burden.14 Furthermore, as the incidence of premature births worldwide increases and as medical technology becomes better able to treat the complications of premature birth, the number of infants at risk for ROP is increasing rapidly.58

Oxygen plays a central role in ROP.913 Oxygen environment and a key transcription factor that oxygen regulates (e.g. Hypoxia inducible factor [HIF]) are thought to modulate ROP. In terms of ROP pathogenesis, a two-phase hypothesis has been proposed and has become widely accepted.14,15 In phase 1, there is delayed physiologic retinal vascular development and vasoattenuation, which is aggravated by hyperoxia and loss of nutrients and growth factors. In phase 2, vasoproliferation occurs at the junction of vascularized and avascular retina. Mouse oxygen-induced retinopathy (OIR) model (exposure to 75% oxygen for 5 days followed by room air), a widely used animal model of ROP, best represents the two-phase hypothesis.16,17 During the vasoproliferative phase, the avascular retina releases pro-angiogenic growth factors such as vascular endothelial growth factor (VEGF), which are induced by hypoxia and may cause aberrant vessel growth and neovascularization. Oxygen fluctuations with intermittent hypoxia is also implicated in development of ROP in clinical studies1820 and OIR animal model studies especially in rats (e.g. cycling between 50 and 10% oxygen).21,22 Growing neovascular vessels lead to fibrovascular membranes that may pull on the retina, causing tractional retinal detachment and eventual blindness. The phenotype of ROP is classified based on location, extent, and severity of these pathologic changes.23 Some infants show a rapidly progressing, severe form of ROP, known as aggressive posterior ROP (AP-ROP).2327

Early investigations into ROP risk factors focused primarily on prematurity itself, as well as environmental factors including oxygen exposure after birth.10,11 Various studies focusing on oxygen exposure have proven its importance as a primary predictor of ROP outcomes.911 However, some high-risk infants with extremely low birth weight (BW) and gestational age (GA) do not develop ROP, whereas some low-risk infants do develop severe ROP. In these infants at phenotypic extremes, a study showed that known clinical risk factors were not significantly associated with development of ROP.28 In addition, it is not understood why certain infants are predisposed to AP-ROP with very high likelihood of blindness. This heterogeneity of ROP risk suggests that other factors, such as genetics may be involved in creating a predisposition to ROP. Before specific genetic variations were investigated in ROP, epidemiologic studies suggested racial and ethnic differences in ROP incidence.2931 The Cryotherapy for ROP (CRYO-ROP) study of 4,099 premature infants found 7.4% of white infants reached threshold disease, while only 3.2% of black infants achieved a similar level of disease.31 Also, twin and sibling studies have supported the involvement of a genetic component of disease. Two studies of monozygotic and dizygotic twins found that the heritability of ROP was 0.70 and 0.73, respectively.32,33 Evidence of genetic effects is also supported by data from the oxygen-induced retinopathy (OIR) phenotype in rodent models, in which studies of different rat strains have found differences in the retinal avascular area and VEGF expression between strains.3436 Investigations into this genetic component in humans and animal models have implicated the involvement of multiple genes, but have not discovered a genetic component of large effect. It is likely that knowledge of such a genetic component could be used to identify possible targets to improve outcomes of screening and treatment.

Many signaling molecules and related pathways have been suspected in the pathogenesis of ROP due to known biochemical and clinical associations: VEGF, insulin-like growth factor-1 (IGF-1), erythropoietin (EPO), and inflammatory mediators. In addition to ROP, the growth of abnormal, leaky blood vessels is a common pathologic component of other blinding neovascular eye diseases, such as diabetic retinopathy (DR) and neovascular age-related macular degeneration (AMD), both of which have strong evidence of a genetic predisposition to disease.3739 Moreover, because ROP progresses more rapidly and presents with relatively homogeneous clinical characteristics, the correlation of genotype and phenotype is easier than with a chronic disease such as DR or AMD.15 Thus, the study of ROP genetics may give us important insights into the pathophysiology of other more prevalent adult and pediatric neovascular retinal diseases.

This review summarizes current research into genetic factors contributing to ROP risk in both human and animal models and recommends future directions for research into the underlying genetics of pathways that contribute to disease.

METHODS

Pubmed was queried from January 1980 to June 2017. The following search terms were used: retinopathy of prematurity AND genetics, retinopathy of prematurity AND gene, retinopathy of prematurity AND single nucleotide polymorphism (SNP), retinopathy of prematurity AND variant, and retinopathy of prematurity AND polymorphism. Criteria for inclusion included the relevance, clinical importance, level of statistical evidence provided, and scientific importance of articles to the subject of this paper. Articles cited in the reference lists of other articles were reviewed and included when considered appropriate. All articles with English abstracts were reviewed.

CANDIDATE GENES IN ROP

VEGF and associated receptors

VEGF plays a crucial role in ROP. Increased VEGF in avascular retina stimulates pathological retinal neovascularization, which may result in blinding complications like tractional retinal detachment. Moreover, VEGF is a proven therapeutic target, as intravitreal anti-VEGF therapy has shown efficacy in promoting regression of severe ROP.40 There have been many genetic studies on associations between the VEGF gene and incidence or severity of ROP.

Table 1 summarizes results of SNP studies in human VEGF gene (VEGFA). rs2010963 (also known as −634G>C and +405 G>C) is the most extensively studied SNP. In a British study of 188 preterm infants on rs2010963 in 2004, the G allele was found to have higher frequency among infants with ROP.41 This result was supported by a 2015 study in 102 preterm infants from Egyptian hospitals showing that G allele was significantly higher in infants with ROP.42 However, one study in Hungary reported the opposite results – higher frequency of C allele in severe ROP – and 5 other studies found no significant association between rs2010963 and ROP.

Table 1.

Studies Investigating the Association Between VEGFA Genes and Retinopathy of Prematurity (ROP)

Polymorphism Study country Subjects Results Reference
rs2010963
(−634G>C, +405G>C)
United Kingdom 91 treatment-requiring ROP (BW 779g [440–1185g], GA 25 wk [23–32 wk]), 97 non-treatment-requiring preterm infants (BW 920 g [448–2302g], GA 26 wk ± 2.9 wk) Higher frequency G allele among infants with threshold ROP 41
Hungary 115 treatment-requiring ROP (BW 1160g ± 270g, GA 28.5 wk ± 2.0 wk), 86 mild or no ROP (BW 1200g ±270g, GA 29.2 wk ± 2.9 wk) Higher frequency C allele among treated infants 137
Turkey 42 treatment-requiring ROP (BW 1097.5g ± 264.3g, GA 28.2 wk ± 2.4 wk), 50 regressed ROP (BW 1253.0g ± 212.2g, GA 29.7 wk ± 2.0 wk), 31 no ROP (BW 1345.6g ± 225.9g) No significant association 138
United States 61 stage 4/5 ROP (BW 882g [600–1300g], GA 26 wk [23–30 wk]), 61 normal controls (BW 2430–3960g, GA 34–40 wk) No significant association 139
Japan 127 ROP (944g [3778–2168g], GA 27 wk [22–33 wk]), 77 no ROP (BW 1596g [692–2400g], GA 32 wk [22–33 wk]) No significant association 140
Egypt 62 ROP (BW 1400g [1000–2110g], GA 32 wk [28–34 wk]), 40 no ROP (BW 1640g [1009–2800g], GA 33 wk [29–35 wk]) High frequency of G allele in ROP 42
Poland 60 treatment-requiring ROP (BW 900g ± 225g, GA 26.7 wk ± 2.3 wk), 20 regressed ROP (BW 1029g ±231g, GA 27.5 wk ± 1.6 wk), 101 no ROP (BW 1153g ±225g, GA 29.2 wk ± 2.05 wk) No significant association 141
Iran 15 treatment-requiring ROP (BW 879g ± 81g, GA 27 wk ± 13 wk), 30 regressed ROP (BW 884g ± 63g, GA 27 wk ± 12 wk), 66 no ROP (BW 980g ± 81 g, GA 27 wk ± 10 wk) No significant association 142
rs1547651 Caucasian 43 ROP, 299 no ROP (all subjects GA ≤ 28 weeks) No significant association 43
rs3025039
(+936C>T)
Caucasian 43 ROP, 299 no ROP (all subjects GA ≤ 28 weeks) No significant association 43
Iran 15 treatment-requiring ROP (BW 879g ± 81g, GA 27 wk ± 13 wk), 30 regressed ROP (BW 884g ± 63g, GA 27 wk ± 12 wk), 66 no ROP (BW 980g ± 81 g, GA 27 wk ± 10 wk) No significant association 142
United States 33 stage 4/5 ROP, 49 normal controls No significant association 143
Egypt 62 ROP (BW 1400g [1000–2110g], GA 32 wk [28–34 wk]), 40 no ROP (BW 1640g [1009–2800g], GA 33 wk [29–35 wk]) No significant association 42
rs833058 Italy 43 ROP, 299 no ROP (all subjects GA ≤ 28 weeks) No significant association 43
rs833061
(−460C>T)
Italy 43 ROP, 299 no ROP (all subjects GA ≤ 28 weeks) No significant association 43
Hungary 115 treatment-requiring ROP (BW 1160g ± 270g, GA 28.5 wk ± 2.0 wk), 86 mild or no ROP (BW 1200g ± 270g, GA 29.2 wk ± 2.9 wk) High frequency of 460TT/405CC haplotype in treatment-requiring ROP 137
Turkey 42 treatment-requiring ROP (BW 1097.5g ± 270g, GA 28.2 wk ± 2.4 wk), 50 regressed ROP (BW 1253.0g ± 212.2g, GA 29.7 wk ± 2.0 wk), 31 no ROP (BW 1345.6g ± 225.9g) No significant association 138
United States 61 stage 4/5 ROP (BW 882g [600–1300g], GA 26 wk [23–30 wk]), 61 normal controls (BW 2430–3960g, GA 34–40 wk) No significant association 139
+13553C>T Japanese 127 ROP (BW 944g [378–2168g], GA 27 wk [22–33 wk]), 77 no ROP (BW 1596g [692–2400g], GA 32 wk [22–33 wk]) A significant association between the TT genotype and non-severe ROP for gestational age 140
+702C>T United States 33 stage 4/5 ROP, 49 normal controls No significant association 143
+1612G>A United States 33 stage 4/5 ROP, 49 normal controls No significant association 143
−2578C>A United States ROP (BW 2430–3960g, GA 34–40 wk), no ROP (BW 600–1300g, GA 23–30 wk) (number of patients not reported) No significant association 144
Hungary 90 treatment-requiring ROP (BW 1160g ± 300g, GA 28.5 wk ± 2.4 wk), 110 mild (stage 1 or 2) or no ROP (BW 1200g ± 280g, GA 28.5 wk ± 2.4 wk) No significant association 145

Table lists investigated polymorphism and presence of statistical significance. Where noted in the original study, information is provided in parentheses regarding the birth weight (BW) and gestational age (GA) of patients. Brackets denote range of patient values and ± denotes one standard deviation of range of each variable.

In addition, rs833061 (−460C>T) and VEGFA +13553C>T have been reported to be associated with ROP. However, replication has not been attempted for +13553C>T and the association of rs833061 and ROP has not been replicated in 3 other studies. VEGFA haplotypes have also been reported to be associated with ROP. A study performed in an Italian population of 342 infants focused on the distribution of polymorphisms in a handful of genes implicated in ROP showed evidence that VEGFA haplotype (TCCT) decreases risk of ROP.43

VEGF promotes angiogenesis and hyper-permeability by binding to the VEGF receptor 2 (VEGFR-2) on vascular endothelium, whereas VEGFR-1 acts as a decoy receptor.44 However, studies on VEGFR-1 (FLT1) and -2 (KDR) genes found no associations with ROP (Table 2).

Table 2.

Summary of candidate gene studies of retinopathy of prematurity other than VEGFA

Gene Variant Study country Subjects Results Reference
ACE rs1799752 Italy 299 ROP, 43 no ROP No significant association 43
rs4291
287-bp insertion in intron 16 Kuwait 74 ROP (53 regressed, 21 stage 4/5 ROP), 107 no ROP The incidence of the II genotype was higher in ROP cases, while the incidence of the DD genotype was significantly higher in advanced stage ROP cases compared to spontaneously regressing ROP cases. (I, insertion, D, deletion) 90
AGT rs699 Italy 43 ROP, 299 no ROP No significant association 43
AGTR1 rs5186 Italy 43 ROP, 299 no ROP No significant association 43
rs427832 United States 102 ROP, 228 no ROP Significant association with ROP at p < 0.01 level of significance 91
ANGPT2 −35G>C United States Not specified No significant association 144
Hungary 90 treatment-requiring ROP, 110 no or mild (stage 1 or 2) ROP No significant association 145
BDNF rs7934165
rs2049046
United States 126 treatment-requiring ROP, 467 stage 1/2 ROP Two intronic SNPs found to be associated with difference between mild and threshold ROP 146
rs7934165 United States 140 treatment-requiring ROP, 1257 no or mild (stage 1 or 2) ROP Meta-analysis of two studies provided evidence of association of variant with severe ROP 146
CETP rs289747 United States 102 ROP, 228 no ROP Significant association with ROP at p < 0.01 level 91
CFH rs52985 United States 102 ROP, 228 no ROP Increased protection against ROP as number of T alleles increased (p = 0.01) 91
rs800292 United States 102 ROP, 228 no ROP Increased protection against ROP as number of T alleles increased (p = 0.01) 91
EPAS1 rs1867785 United States 102 ROP, 228 no ROP Significantly higher incidence of A allele in ROP 91
GP1BA rs2243093 United States 102 ROP, 228 no ROP Significant association with ROP at p < 0.01 level 91
LRP5 rs143924910
(c.3656G>A),
c.4148A>C,
rs141407040
(c.4619C>T)
Japan 53 advanced ROP Direct sequencing of coding regions of LRP5 revealed 3 nonsynonymous DNA variants in 3 patients. 56
3-bp insertion in exon 1 Japan 17 advanced ROP, 51 no ROP Single patient with advanced ROP shown to have 3 bp insertion in exon 1 CTG repeat area not observed in 28 unaffected patients. 147
NOS3 rs2070744
(−786T>C)
Italy 43 ROP, 299 no ROP No significant association 43
Hungary 105 treatment-requiring ROP, 127 stage 1 or 2 ROP No significant association 148
United States 15 ROP, 131 no ROP significantly higher frequency of C allele in ROP 149
United States 19 stage 4/5 ROP, 34 normal significantly higher frequency of C allele in ROP 150
rs1799983
(894G>T)
United States 14 stage 4/5 ROP, 32 normal No significant association 150
United States 15 ROP, 131 no ROP significantly higher frequency of T allele in ROP 149
Italy 43 ROP, 299 no ROP No significant association 43
27-bp VNTR in intron 4 (b/a) United States 15 stage 4/5, 32 normal controls No significant association 150
Hungary 105 treatment-requiring ROP, 127 stage 1 or 2 ROP The aa genotype presented an independent risk factor for ROP requiring treatment. 148
rs61722009 Italy 43 ROP, 299 no ROP No significant association 43
FLT1 c.+6724(TG) 13–23 dinucleotide repeat Japan 127 ROP, 77 no ROP No significant association 140
FZD4 c.97 C>T; c.502 C>T
(double missense mutation)
United States 93 ROP, 98 normal controls Seven of 93 (7.5%) patients with ROP showed c.97 C>T; c.502 C>T double missense mutation. 57
rs80358282
(c.205C>T),
rs184709254
(c.380G>A),
c.631T>C
Japan 53 advanced ROP Direct sequencing of coding regions of FZD4 revealed 3 nonsynonymous DNA variants in 4 patients. 56
c.766A>G Unspecified 10 sporadic FEVR cases
20 advanced ROP cases
PCR amplification of a large DNA fragment revealed one severe ROP case with c.766A>G. Significance not investigated. 54
c.1109C>G,
c.609G>T
Canada 71 severe ROP, 33 mild or no ROP Direct sequencing of coding regions of FZD4 revealed 2 nonsynonymous DNA variants in 2 patients. 55
HMOX1 rs3074372 Italy 43 ROP, 299 no ROP No significant association 43
IGF1R c.3174G>A United States 52 stage 4/5 ROP, 33 normal controls No significant association 71
Hungary 108 treatment-requiring ROP, 120 stage 1 or 2 ROP, 164 normal controls No significant association 70
IHH rs3099 United States 102 ROP, 228 no ROP Significant association with ROP at p < 0.01 level 91
IL10 −1082G>A Germany 31 stage 1 or 2 ROP, 13 stage 3 ROP, 29 no ROP No significant association 76
IL1B +3953C>T Germany 31 stage 1 or 2 ROP, 13 stage 3 ROP, 29 no ROP No significant association 76
KDR 32G>A Turkey 42 treatment-requiring ROP, 50 regressed ROP, 31 normal controls No significant association 138
g.+4422(AC)11–14 dinucleotide repeat Japan 127 ROP, 77 no ROP No significant association 140
NDP Sequencing of all 3 exons and UTRs United States 54 severe ROP, 36 mild or no ROP, 22 normal controls, 31 normal parents Six of 54 (11 %) infants with severe ROP had polymorphisms in the NDP. 151
Direct sequencing of coding regions and noncoding exon 1 Japan 53 advanced ROP No meaningful sequence changes 56
237A>G Japan 17 advanced ROP, 51 no ROP Single patient with AP-ROP found to have heterozygous substitution not observed in 51 unaffected cases 147
14 bp deletion in exon 1 Australia 31 ROP (Stage 2 or greater), 90 no ROP Two twins with stage 3 regressed ROP and one unrelated patient with regressed stage 2 ROP displayed 14 bp deletion in CT repeat reagion. Also observed in a control patient. No statistical analysis. 152
5 bp deletion in exon 1
26C>G
71 bp deletion in exon 1
UK 31 ROP stage 3 or more, 16 regressed ROP, 2 no ROP One patient had 5 bp deletion and C>G transersion at +26, one patient had 71 bp deletion in same exon 1 region. No statistical analysis. 61
12 bp insertion in exon 1
14 bp deletion in exon 1
Japan 100 advanced ROP (stage 4/5), 6 regressed stage 3 ROP, 130 no ROP Two advanced ROP patients found to have disruptions in exon 1 of ND gene. No statistical analysis. 59
597C>A
110C>G
Kuwait 95 ROP, 115 no ROP Significant association was found between ROP and 597C>A polymorphism. No significance found between 110C>G polymorphism and ROP. 60
121C>T
R121W
L108P
United States 16 ROP, 50 normal controls One patient with a heterozygous base substitution, one pair of twins with novel R121W mutation, and one pair of twins with L108P missense mutation observed. No statistical analysis. 58
TBX5 rs1895602 United States 102 ROP, 228 no ROP Significant association with ROP at p < 0.01 level 91
TGFB1 −509C>T United Kingdom 91 treatment-requiring ROP, 97 stage 1/2 or no ROP No significant association 41
TLR4 rs4986790
(c.896A>G)
Germany 31 stage 1 or 2 ROP, 13 stage 3 ROP, 29 no ROP No significant association 76
TNF −308G>A United Kingdom 91 treatment-requiring ROP and 97 stage 1/2 or no ROP No significant association 41
Germany 31 stage 1 or 2 ROP, 13 stage 3 ROP, 29 no ROP No significant association 76
TSPAN12 Direct sequencing of coding regions of TSPAN12 Japan 53 advanced ROP No meaningful sequence changes 56

FEVR, Norrie disease and the Wnt pathway

Familial Exudative Vitreoretinopathy (FEVR) and Norrie disease are developmental diseases of the retina with known genetic causes with similar pathology to ROP. Both are hereditary disorders occurring primarily in full-term infants, characterized by abnormal retinal vascularization leading to retinal detachment.45,46 While patients with Norrie disease are blind from or shortly after birth, and often have systemic pathologies such as deafness and mental retardation, the clinical manifestations of FEVR are variable but restricted to abnormalities in ocular development.47 FEVR is known to be caused by mutations in FZD4, LRP5, TSPAN12, NDP, etc.,4851 and Norrie disease is caused by mutations in the NDP gene.46 These genes encode proteins which are components of the Wnt/beta-catenin signaling pathway – a group of signal transduction pathways with roles in cell survival, proliferation, and migration throughout the body.

The canonical (beta-catenin dependent) Wnt pathway has known roles in a variety of diseases with angiogenic properties including DR and AMD.52,53 Frizzled-4 and low-density-lipoprotein receptor related protein 5 (LRP-5) are receptors for Wnt ligands, and tetraspanin-12 is an auxillary membrane protein. Norrin, a product of NDP gene, binds to the Frizzled-4, LRP-5, and tetraspanin-12 receptor complex and activates signals on endothelial cells. Mutations of these genes have been investigated in ROP (Table 2).

Mutations in the FZD4 gene were found in up to 7.5% of patients with severe ROP (Table 2).5457 A 2015 study of 421 patients displaying various vitreoretinopathies found a significant association between the FZD4 double missense mutation [P33S(;)P168S] and both ROP and FEVR.57 A study of 53 Japanese patients with advanced ROP was performed using direct sequencing of FZD4, TSPAN12, NDP, and LRP5. Investigators identified six nonsynonymous DNA variants in the coding regions of FZD4 and LRP5, but detected no changes in NDP or TSPAN12, demonstrating involvement of Wnt with ROP.56

Mutations in the NDP gene have also been found in ROP patients with variable frequencies (Table 2).5860 SNP studies in Kuwaiti populations have supported evidence of a link between NDP and ROP,60 while other studies have implied that mutations in the regulatory region of NDP are also a contributor to the development of ROP.61 The relationship between SNPs residing in the UTR of NDP and progression of ROP to advanced disease has also been investigated. The Kuwaiti study by Haider found that 83% of patients with severe disease possessed NDP 597C>A polymorphisms in their UTR, while none of those whose disease resolved spontaneously possessed this polymorphism.60

Taken together, these findings intriguingly suggest involvement for the Wnt pathway and associated genes in ROP development, and serve as strong candidates for further sequencing research. It should be noted that it may be difficult or nearly impossible to differentiate ROP from FEVR in premature infants. This has recently been proposed as a new classification, ROPER (ROP vs. FEVR) due to the clinical similarity of the two conditions.62 In future studies, in-depth analysis of clinical features, retinal imaging with fluorescein angiography, genetic and phenotypic analysis of relatives, and functional analysis of genetic variants may be helpful for better understanding of genetics in ROP as well as FEVR.

IGF-1

Insulin-like growth factor 1 (IGF-1), a growth hormone promoting somatic growth and maturation, has also been proposed as a contributing factor to ROP progression.63 IGF-1-deficient mice showed a decrease in vascular development61 and lower birth weight64 than those of controls. In human babies, low IGF-1 levels were also associated with low birth weight,65 and persistent low serum IGF-1 levels were associated with severity of ROP.63,66 Based on these findings, IGF-1 replacement therapy has recently been investigated.67 A phase 2 trial of administering a complex of recombinant human IGF-1 and IGFBP-3 to prevent ROP was undertaken, but the study did not meet its primary endpoint of reducing severity of ROP.68

Investigations of specific polymorphisms of IGF-1 gene have been unsuccessful finding a significant association. A study linked a c.3174G>A polymorphism in the IGF-1 receptor gene (IGF1R) to low levels of plasma IGF-1.69 A 2006 study of 392 infants in Hungary was unable to detect a difference in the prevalence of the IGF1R c.3174G>A among severe ROP, mild ROP and full-term groups (Table 2).70 A 2007 study in an American population was also unable to find a link between advanced ROP and IGF1R c.3174G>A polymorphism (Table 2).71

eNOS

Endothelial nitric oxide synthase (eNOS) is one of the constitutive enzymes that synthesize NO, which is known to play a regulatory role in retinal and choroidal blood flow.72,73 In an eNOS-deficient mouse OIR model, neovascularization and vaso-obliteration were both reduced.74 Moreover, eNOS gene polymorphisms have shown reduced NO levels.75 Thus, the association between ROP and eNOS gene (NOS3) polymorphisms have been investigated. A literature search showed that 3 SNPs (rs2070744, rs1799983 and rs61722009) and one variable number tandem repeat (VNTR), 27-bp VNTR in intron 4, had been observed in ROP patients (Table 2). Although some studies reported positive associations between rs2070744, rs1799983, or the 27-bp VNTR and ROP, others found contradictory results (Table 2).

Inflammatory Mediators

Growing evidence suggests that perinatal inflammation and infection may increase the risk for ROP by direct proangiogenic effects and/or modifying known risk factors.76 Studies have reported higher plasma levels of inflammatory cytokines including IL-6, Il-8, and TNF77 and higher vitreous levels of inflammatory cytokines including IL-6, IL-7, IL-10, IL-15, etc. in eyes with advanced ROP.78

Dammann et al investigated 4 SNPs of inflammation-associated genes (IL1B, TNF, IL10, TLR4) in preterm patients, but none showed significant association, although there were trends towards higher stage of ROP with the presence of TNF and IL1B SNPs (Table 2).76 TNF −308G>A polymorphism also showed no significant associations with ROP (Table 2).

A recent study has also shown an angiogenic role for mast cells and associated factors including mast cell tryptase and monocyte chemotactic protein-1, making them a potential target for ROP research.79

Brain-derived neurotrophic factor

Brain-derived neurotrophic factor (BDNF), a neuronal trophic factor in brain and retina, may promote survival of several types of retinal neurons.8083 Although the exact role of BDNF in retinal angiogenesis is unknown, reduced BDNF levels have been demonstrated in patients with severe ROP, suggesting a possible role of BDNF in development of severe ROP.8486 In an animal model study, the retinal level of BDNF was lower in the OIR mouse model compared to that in normal controls.86

In a large-scale candidate gene study, which analyzed 1614 Tag SNPs of the 145 candidate genes in 817 infants in the discovery cohort and 543 in the US replication cohort, it was found that two SNPs (rs7934165 and rs2049046) in the intronic region of BDNF were associated with severe ROP. Although these results were not independently confirmed in the replication cohort, the association with rs7934165 did increase in significance with severe ROP in their meta-analysis of the combined data. Interestingly, reduced serum BDNF in the severe ROP group was also found in the same discovery cohort.87 Further studies on the functional effects of intronic variants of BDNF and replication studies in different populations are warranted.

Renin-Angiotensin System

The Renin Angiotensin system (RAS) has been linked to retinal vascular development and pathological angiogenesis. Blockade of RAS with inhibitors of angiotensin-converting enzyme (ACE) and angiotensin receptor blockers ameliorated OIR, suggesting that inhibiting RAS may be beneficial in ROP.88 A SNP study of ACE gene showed association with DR.87

However, results from genetic studies on RAS component genes in ROP are inconclusive (Table 2). A study in Italy showed no associations between ROP and SNPs of ACE gene (ACE), angiotensinogen gene (AGT) and angiotensinogen type 1 receptor gene (AGTR1). In a study of 181 premature Kuwaiti infants on 287-bp insertion(I)/deletion(D) in intron 16, the frequency of II genotype was higher in ROP patients compared to normal controls, but the frequency of DD genotype was higher in advanced ROP patients compared to regressed ROP.90 A candidate gene study of 228 infants with ROP and 102 controls found a SNP in the AGTR1 gene to be associated with ROP, though this association was not significant after Bonferroni correction.91

Angiopoietins

Angiopoietin(Ang)-1 and -2 are growth factors that are essential for retinal vascular development. Ang-1 binds tyrosine kinase receptor Tie2 and promotes vascular maturation and stabilization.92 In an OIR model, intravitreal Ang-1 promoted normal vascular regeneration while inhibiting pathological angiogenesis and vascular leakage.93 In contrast, Ang-2, a competitive antagonist of Ang-1/Tie-2, promotes neovascularization in animal models.94,95 Vitreous levels of Ang-1 and Ang-2 in eyes of stage 4 ROP were higher than those of control eyes.96 However, in two studies of Ang-2 gene promoter polymorphism (ANGPT2 −35G>C), no association was found with ROP (Table 2).

Erythropoietin

Erythropoietin (EPO), a hormone known to stimulate red blood cell formation in bone marrow, and EPO receptors are expressed in retina, and their expression is regulated by oxygen status.97,98 Mouse models of ROP have shown that vascular stability is affected by EPO levels, with exogenous restoration of EPO leading to a reduction in blood vessel dropout during the first phase of ROP.96 Conversely, elevated levels of EPO during the second stage of ROP exacerbated vasoproliferation, and the vitreous level of EPO is elevated in eyes with stage 4 ROP. Increased erythropoietin receptor signaling has also been shown to influence severe OIR models of disease through VEGFR2-mediated angiogenesis, making it an important target for clinical research in human patients.99,100 While a variant of EPO was investigated in a candidate-gene study by Mohamed et. al., significance for this variant was not reported in the study results.91

Hypoxia inducible factor

HIF-1 plays a central role in oxygen homeostasis.101 According to the oxygen environment, HIF-1 regulates transcription of genes such as VEGF, VEGFR1, PDGF, SDF-1 and Ang2, which have been suggested to play important roles in retinal angiogenesis.94 In a study of Hif1α knockout mice in an OIR model of disease, disruption of HIF-1 was shown to lead to decreased VEGF abundance, indicating a possible role in neovascularization.102 Additionally, organ system pharmacology studies in mouse models have indicated that stabilization of HIF-1 may be important for protection against oxygen toxicity in premature infants.103

Likewise, homologous recombination models in mice studying HIF-1a-like factor (HLG) and HIF2α found decreasing expression of these genes led to decreased EPO expression and resistance to hyperoxia treatments meant to induce ROP.104 HIF1α was also shown to upregulate annexin A2 expression in OIR mice during hypoxia, supporting a role in OIR models.105

HIF2α’s closest human analogue, known as Endothelial PAS Domain Protein 1 (EPAS1), serves as the main regulator of EPO induction and has also been shown to have a connection to ROP.106 A candidate gene study of 153 genes in 347 infants under 32 weeks gestational age found an association between EPAS1 with development of severe ROP.91

Heme oxygenase-1

Heme oxygenase-1 plays important roles in inflammatory responses, oxidative stress, iron-metabolism, and vascular physiology. However, in a candidate gene study, rs3074372 in HMOX1 showed no significant association with ROP (Table 2).

Other candidate factors

In addition to the above described factors and pathways, a number of other potential targets and mechanisms have been identified that lack genetic studies in patients with ROP. The ‘a’ disintegrin and metalloproteinase (ADAM) family of proteases are involved in the degradation of extracellular matrix components as well as interactions mediated by integrin.107 Several subtypes of ADAM family are implicated in the pathogenesis of ROP. ADAM17 knockout mice showed less neovascularization in OIR models without affecting normal vascular development.108 Moreover, ADAM 8, 9, and 10 was found to play a role in development of plus disease in OIR mouse models. Adam8−/− and Adam9−/− mice and mice lacking ADAM10 in endothelial cells showed less severe tortuosity and dilation mimicking less plus disease in ROP.109 Further evaluations in humans including genetic analysis are warranted.

In conjunction with ADAM17, studies have also considered the family of tissue inhibitor of metalloproteinases (TIMP) family of proteins. The TIMP-3 protein specifically is a known physiological ADAM17 inhibitor.110 Mouse model investigations into the application of this protein as a potential treatment showed that TIMP-3 application was linked to decreased neovascular tuft formation.109

In addition to these studies, large candidate gene studies of ROP have been successful identifying targets with undiscovered connections to ROP. The previously mentioned study by Mohamed et al. implicated genes with function in embryonic development (IHH), transcription (TBX5), and protein localization (GP1BA, CETP) (Table 2).91 The same study also found an association between ROP and complement factor H (CFH), known to be associated with development of AMD.38

DISCUSSION

Summary of previous studies

Most genetic studies in ROP have used the candidate gene approach and focused on genes related to angiogenesis, inflammation, and retinal (neuro)development. Among them, VEGFA polymorphisms and FEVR-related genes have been most extensively studied in different populations. However, no VEGFA polymorphisms have been proven to be associated with ROP, because most positive studies have not been replicated in other populations (Table 1). Variants of Wnt pathway genes, which are known to cause FEVR or Norrie disease, have been also found in ROP patients, suggesting possible associations of these variants in at least a small proportion of severe ROP patients (Table 2). However, these results also have limitations in that we may not confidently distinguish between premature infants with severe ROP and FEVR-related genetic variants and prematurely-born infants with FEVR, as Hartnett et al. pointed out.27 In addition the polygenic nature of many diseases makes identification of causative variants difficult in small sample sizes focused on a small number of variants.111 Recently, results of a large-scale candidate gene study using Tag SNPs of the 145 candidate genes in a multiracial cohort were reported.87 Although no SNPs were significantly associated with the presence versus absence of ROP in this study, one SNP of BDNF gene was significantly associated with severe ROP in their meta-analysis combining the discovery and replication cohorts, which warrants further genetic and biological studies.

Limitations of previous studies

It is difficult to draw meaningful conclusions from most of the candidate gene studies reviewed here due to the following limitations: (1) the sample sizes of most individual studies were small; (2) no replication study has been performed for many variants; (3) there are conflicting results among studies of the same variants; (4) most studies were conducted using only one or a few clinical sites; (5) ocular phenotype was not standardized; (6) confounding variables were not reported or standardized; (7) meta-analysis is not possible for most variants due to different study protocols between studies; (8) there are variabilities in neonatal care such as oxygen treatment protocol9, incidence of (severe) ROP, and diagnosis and management of ROP between physicians, study hospitals, study countries and study periods.8,112115 Differences in neonatal care may affect survival rate, systemic morbidities of prematurity, incidence of ROP and severity of ROP, making it difficult to find exact roles of genetic variants. Moreover, there are unexplained differences in outcome of premature birth such as mortality. Also, differences in diagnosis and management of ROP may cause bias in phenotypic categorization of subjects, which is a huge problem in genetic studies. It should be noted that genetic risk factors for stage 1-3 ROP and stage 4 or 5 ROP could be different, as different biochemical processes may be involved and management protocols and treatment outcomes of study centers are also important factors for stage 4 or 5 ROP.

Most importantly, candidate gene studies have inherent limitations of not being able to find novel genetic factors. Other approaches to detect novel variants or genes associated with ROP are necessary.

Future Directions of studying ROP genetics

It is very challenging to study the genetics of multifactorial diseases such as ROP. To overcome the current limitations mentioned above and to study the contribution of genetics efficiently, it is necessary to improve the methodology for studying the genetics of ROP. It is essential that investigators leverage new methods that interrogate genetic factors agnostically and at high sample sizes, in order to maximize study power and facilitate simultaneous investigation of many, rather than single, genetic elements. Genome-wide Association Studies (GWAS) test for association across hundreds of thousands of SNPs simultaneously using array-based technology. GWAS can be helpful to find genes or pathways associated with ROP. In other ophthalmological diseases such as AMD38,116118, DR119,120, glaucoma121123 and myopia124126, GWAS has been successful in finding susceptibility loci. However, a large-scale GWAS has not been conducted in ROP. Massively parallel sequencing, also called next-generation sequencing (NGS), enables sequencing of specific regions, whole exome, or whole genome in a short period of time at high depth and affordable cost. Whole exome sequencing or targeted exome sequencing can be helpful for finding novel variants with possible functional consequences. Exome genotyping arrays may also provide a method of interrogating for SNPs involved in ROP.

In addition to these genetic evaluations, integration of sequence data with data regarding post-transcriptional and post-translational modification, including transcriptomics, metabolomics, and proteomics, will be important to identify biomarkers that may be useful for early detection, diagnosis, and prediction of treatment response. Studies of epigenetics in DR have also shown promise, with epigenetic changes associated with processes of microvasculature complications127, mitochondrial dysfunction128, microRNA expression129, and capillary cell apoptosis.130,131 These findings suggest that interrogation of epigenetic factors may be an important method of discovering new treatments in ROP.

Second, large-scale multi-center collaboration of the type offered by consortium studies can help provide structure to such studies. Consortium approaches facilitate recruitment of larger cohorts and make available more sophisticated computational approaches allowing investigators to control for more complicated confounding effects. Previous large international consortium attempts at examining the role of genetics in multifactorial disease have met with success38,121,132134, and two consortium studies investigating the genetic causes of ROP are currently ongoing at centers in North America.135,136

Third, standardization of ocular phenotypes and confounding factors is crucial. For this, ocular and systemic factors should be acquired systematically, and known risk factors including GA and BW should be assessed in a standardized fashion and strictly controlled for. Additionally, the importance of environmental effects should be noted, as differences between study populations and sites has the ability to have a profound effect on phenotype. Heterogeneity of study subjects in race, ethnicity, and physical covariates, as well as differences between treatment sites and attending clinicians can affect study outcomes. This is especially important to distinguish genetic variants associated with ROP from those associated with prematurity itself. Also, objective phenotyping such as image-based diagnosis should be considered. Compared to clinical ophthalmoscopic diagnosis, consensus image-based diagnosis may enable reduction of intra- and inter-grader discrepancy in ROP diagnosis.

It is also important to note that additional basic research studies using representative animal models such as mouse or rat OIR models are required to test hypotheses. While animal models face many limitations including differences in biology, most notably their use of full-term rather than premature animals, these models’ ability to control for phenotypic, environmental, and genetic stratification factors distinguishes them as a valuable method of testing hypotheses and adding insight to human observational studies.

Expected benefits of genetic studies of ROP

Finding genetic variants affecting ROP will be useful in at least three ways. First, genetic risk factors may be incorporated into risk modelling to predict development and progression of ROP. A refined risk analysis system with clinical and genetic risk factors may help clinicians to identify both high- and low-risk patients. Second, identifying specific genes or biological pathways that contribute the pathogenesis of ROP may be helpful for development of new therapeutics. In AMD, genetic studies have revealed the importance of complement pathway in the pathogenesis of AMD, which has led to development of new investigational agents under clinical trials such as lampalizumab, an inhibitor of complement factor D. Third, studying ROP genetics can also contribute to the understanding of pathophysiologies of other ocular vascular diseases such as AMD or DR and other angiogenesis-related diseases like cancer.15 Fourth, a better understanding of the genetics of retinopathy of prematurity may lead to better understanding of the pathophysiologic mechanisms of common neonatal diseases of prematurity such as chronic lung disease.

Conclusion

Evidence suggests a genetic contribution to ROP, including epidemiologic studies, twin studies and risk analysis studies. To date, a number of candidate gene studies have been performed. However, it is still unclear which genes or variants are significantly and strongly associated with development and progression of ROP. Large-scale studies using NGS and GWAS with standardized phenotyping have potential to expand understanding of genetic contributions and pathophysiology of ROP.

Supplementary Material

supplement

Acknowledgments

Supported by grants R01EY19474, P30EY10572, and T32EY023211 from the National Eye Institute (Bethesda, MD); by the National Center for Advancing Translational Sciences; by CTSI grant UL1TR001881; by the National Institute of Diabetes and Digestive and Kidney Disease Diabetes Research Center (DRC) grant DK063491; by grant SCH-1622679 from the National Science Foundation (Arlington, VA); and by unrestricted departmental funding from Research to Prevent Blindness (New York, NY).

ABBREVIATIONS

ROP

Retinopathy of Prematurity

AP-ROP

aggressive posterior retinopathy of prematurity

BW

birth weight

GA

gestational age

CRYO-ROP

cryotherapy for ROP

OIR

oxygen induced retinopathy

DR

diabetic retinopathy

AMD

age-related macular degeneration

FEVR

familial exudative vitreoretinopathy

UTR

untranslated region

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

No authors have financial conflicts of interest. Michael F. Chiang is an unpaid member of the Scientific Advisory Board for Clarity Medical Systems (Pleasanton, CA), and a Consultant for Novartis (Basel, Switzerland). R.V. Paul Chan is a member of the scientific advisory board for Visunex Medical Systems (Fremont, CA) and a consultant for Alcon (Fort Worth, TX), Allergan (Irvine, CA), and Bausch and Lomb (St. Louis, MO).

References

  • 1.Reese AB. Symposium: Retrolental fibroplasia (retinopathy of prematurity) Am J Ophthalmol. 1955;40:159. doi: 10.1016/0002-9394(55)91614-7. [DOI] [PubMed] [Google Scholar]
  • 2.Reese A, King M, Owens W. A classification of retrolental fibroplasia. Am J Ophthalmol. 1953;36:1333–1335. [PubMed] [Google Scholar]
  • 3.Tasman W. Vitreoretinal changes in cicatricial retrolental fibroplasia. Trans Am Ophthalmol Soc. 1970;68:548. [PMC free article] [PubMed] [Google Scholar]
  • 4.Aiken JW, Vane JR. Intrarenal prostaglandin release attenuates the renal vasoconstrictor activity of angiotensin. J Pharmacol Exp Ther. 1973;184:678–687. [PubMed] [Google Scholar]
  • 5.Tysinger JW, Weidenthal DT. Nasal heterotopia of the macula in retinopathy of prematurity. Am J Ophthalmol. 1977;83:499–500. doi: 10.1016/0002-9394(77)90555-4. [DOI] [PubMed] [Google Scholar]
  • 6.Tasman W. Retinal detachment in retrolental fibroplasia. Albrecht Von Graefes Arch Klin Exp Ophthalmol. 1975;195:129–139. doi: 10.1007/BF00417115. [DOI] [PubMed] [Google Scholar]
  • 7.Harris G. Retinopathy of prematurity and retinal detachment. Canadian journal of ophthalmology Journal canadien d’ophtalmologie. 1976;11:21–25. [PubMed] [Google Scholar]
  • 8.Gilbert C. Retinopathy of prematurity: a global perspective of the epidemics, population of babies at risk and implications for control. Early Hum Dev. 2008;84:77–82. doi: 10.1016/j.earlhumdev.2007.11.009. [DOI] [PubMed] [Google Scholar]
  • 9.Saugstad OD, Aune D. Optimal oxygenation of extremely low birth weight infants: a meta-analysis and systematic review of the oxygen saturation target studies. Neonatology. 2014;105:55–63. doi: 10.1159/000356561. [DOI] [PubMed] [Google Scholar]
  • 10.Kinsey VE, Jacobus JT, Hemphill F. Retrolental fibroplasia: cooperative study of retrolental fibroplasia and the use of oxygen. AMA archives of ophthalmology. 1956;56:481–543. [PubMed] [Google Scholar]
  • 11.Flynn JT, Bancalari E, Snyder ES, et al. A cohort study of transcutaneous oxygen tension and the incidence and severity of retinopathy of prematurity. N Engl J Med. 1992;326:1050–1054. doi: 10.1056/NEJM199204163261603. [DOI] [PubMed] [Google Scholar]
  • 12.Hartnett ME, Lane RH. Effects of oxygen on the development and severity of retinopathy of prematurity. Journal of American Association for Pediatric Ophthalmology and Strabismus. 2013;17:229–234. doi: 10.1016/j.jaapos.2012.12.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Heidary G, Vanderveen D, Smith LE. Retinopathy of prematurity: current concepts in molecular pathogenesis. Paper presented at: Semin Ophthalmol. 2009 doi: 10.1080/08820530902800314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hartnett ME, Penn JS. Mechanisms and management of retinopathy of prematurity. N Engl J Med. 2012;367:2515–2526. doi: 10.1056/NEJMra1208129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Smith LE. Through the eyes of a child: understanding retinopathy through ROP the Friedenwald lecture. Invest Ophthalmol Vis Sci. 2008;49:5177–5182. doi: 10.1167/iovs.08-2584. [DOI] [PubMed] [Google Scholar]
  • 16.Smith L, Wesolowski E, McLellan A, et al. Oxygen-induced retinopathy in the mouse. Invest Ophthalmol Vis Sci. 1994;35:101–111. [PubMed] [Google Scholar]
  • 17.Connor KM, Krah NM, Dennison RJ, et al. Quantification of oxygen-induced retinopathy in the mouse: a model of vessel loss, vessel regrowth and pathological angiogenesis. Nat Protoc. 2009;4:1565–1573. doi: 10.1038/nprot.2009.187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Cunningham S, Mclntosh N, Fleck B, Elton R. Transcutaneous oxygen levels in retinopathy of prematurity. The Lancet. 1995;346:1464–1465. doi: 10.1016/s0140-6736(95)92475-2. [DOI] [PubMed] [Google Scholar]
  • 19.York JR, Landers S, Kirby RS, Arbogast PG, Penn JS. Arterial oxygen fluctuation and retinopathy of prematurity in very-low-birth-weight infants. J Perinatol. 2004;24:82–87. doi: 10.1038/sj.jp.7211040. [DOI] [PubMed] [Google Scholar]
  • 20.Saito Y, Omoto T, Cho Y, Hatsukawa Y, Fujimura M, Takeuchi T. The progression of retinopathy of prematurity and fluctuation in blood gas tension. Graefe’s archive for clinical and experimental ophthalmology. 1993;231:151–156. doi: 10.1007/BF00920938. [DOI] [PubMed] [Google Scholar]
  • 21.Penn JS, Henry MM, Tolman BL. Exposure to alternating hypoxia and hyperoxia causes severe proliferative retinopathy in the newborn rat. Pediatr Res. 1994;36:724–731. doi: 10.1203/00006450-199412000-00007. [DOI] [PubMed] [Google Scholar]
  • 22.Penn JS, Henry MM, Wall P, Tolman BL. The range of PaO2 variation determines the severity of oxygen-induced retinopathy in newborn rats. Invest Ophthalmol Vis Sci. 1995;36:2063–2070. [PubMed] [Google Scholar]
  • 23.Gole GA, Ells AL, Katz X, et al. The international classification of retinopathy of prematurity revisited. JAMA Ophthalmology. 2005;123:991–999. doi: 10.1001/archopht.123.7.991. [DOI] [PubMed] [Google Scholar]
  • 24.Zacharias L. Retrolental fibroplasia: a survey. Am J Ophthalmol. 1952;35:1426–1454. doi: 10.1016/0002-9394(52)92162-4. [DOI] [PubMed] [Google Scholar]
  • 25.Schaffer DB, Palmer EA, Plotsky DF, et al. Prognostic factors in the natural course of retinopathy of prematurity. Ophthalmology. 1993;100:230–237. doi: 10.1016/s0161-6420(93)31665-9. [DOI] [PubMed] [Google Scholar]
  • 26.Palmer EA. Results of US randomized clinical trial of cryotherapy for ROP (CRYO-ROP) Doc Ophthalmol. 1990;74:245–251. doi: 10.1007/BF02482615. [DOI] [PubMed] [Google Scholar]
  • 27.Hartnett EM. Features associated with surgical outcome in patients with stages 4 and 5 retinopathy of prematurity. Retina. 2003;23:322–329. doi: 10.1097/00006982-200306000-00006. [DOI] [PubMed] [Google Scholar]
  • 28.Port AD, Chan RP, Ostmo S, Choi D, Chiang MF. Risk factors for retinopathy of prematurity: insights from outlier infants. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2014;252:1669–1677. doi: 10.1007/s00417-014-2716-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ng Y, Shaw D, Fielder A, Levene M. Epidemiology of retinopathy of prematurity. The Lancet. 1988;332:1235–1238. doi: 10.1016/s0140-6736(88)90820-3. [DOI] [PubMed] [Google Scholar]
  • 30.Tadesse M, Dhanireddy R, Mittal M, Higgins RD. Race, Candida sepsis, and retinopathy of prematurity. Neonatology. 2002;81:86–90. doi: 10.1159/000047189. [DOI] [PubMed] [Google Scholar]
  • 31.Saunders RA, Donahue ML, Christmann LM, et al. Racial variation in retinopathy of prematurity. Arch Ophthalmol. 1997;115:604–608. doi: 10.1001/archopht.1997.01100150606005. [DOI] [PubMed] [Google Scholar]
  • 32.Bizzarro MJ, Hussain N, Jonsson B, et al. Genetic susceptibility to retinopathy of prematurity. Pediatrics. 2006;118:1858–1863. doi: 10.1542/peds.2006-1088. [DOI] [PubMed] [Google Scholar]
  • 33.Ortega-Molina J, Anaya-Alaminos R, Uberos-Fernández J, et al. Genetic and environmental influences on retinopathy of prematurity. Mediators Inflamm. 2015;2015 doi: 10.1155/2015/764159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.van Wijngaarden P, Coster DJ, Brereton HM, Gibbins IL, Williams KA. Strain-dependent differences in oxygen-induced retinopathy in the inbred rat. Invest Ophthalmol Vis Sci. 2005;46:1445–1452. doi: 10.1167/iovs.04-0708. [DOI] [PubMed] [Google Scholar]
  • 35.Floyd B, Leske DA, Wren S, Mookadam M, Fautsch MP, Holmes JM. Differences between rat strains in models of retinopathy of prematurity. Mol Vis. 2005;11:524–530. [PubMed] [Google Scholar]
  • 36.van Wijngaarden P, Brereton HM, Coster DJ, Williams KA. Genetic influences on susceptibility to oxygen-induced retinopathy. Invest Ophthalmol Vis Sci. 2007;48:1761–1766. doi: 10.1167/iovs.06-0531. [DOI] [PubMed] [Google Scholar]
  • 37.Kuo JZ, Wong TY, Rotter JI. Challenges in elucidating the genetics of diabetic retinopathy. JAMA ophthalmology. 2014;132:96–107. doi: 10.1001/jamaophthalmol.2013.5024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Fritsche LG, Igl W, Bailey JNC, et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat Genet. 2016;48:134. doi: 10.1038/ng.3448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lim LS, Mitchell P, Seddon JM, Holz FG, Wong TY. Age-related macular degeneration. The Lancet. 2012;379:1728–1738. doi: 10.1016/S0140-6736(12)60282-7. [DOI] [PubMed] [Google Scholar]
  • 40.Mintz-Hittner HA, Kennedy KA, Chuang AZ. Efficacy of intravitreal bevacizumab for stage 3+ retinopathy of prematurity. N Engl J Med. 2011;364:603–615. doi: 10.1056/NEJMoa1007374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cooke RW, Drury JA, Mountford R, Clark D. Genetic polymorphisms and retinopathy of prematurity. Invest Ophthalmol Vis Sci. 2004;45:1712–1715. doi: 10.1167/iovs.03-1303. [DOI] [PubMed] [Google Scholar]
  • 42.Ali AA, Hussien NF, Samy RM, Al Husseiny K. Polymorphisms of Vascular Endothelial Growth Factor and Retinopathy of Prematurity. J Pediatr Ophthalmol Strabismus. 2015;52:245–253. doi: 10.3928/01913913-20150506-02. [DOI] [PubMed] [Google Scholar]
  • 43.Poggi C, Giusti B, Gozzini E, et al. Genetic Contributions to the Development of Complications in Preterm Newborns. PLoS One. 2015;10:e0131741. doi: 10.1371/journal.pone.0131741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Miller JW, Le Couter J, Strauss EC, Ferrara N. Vascular endothelial growth factor a in intraocular vascular disease. Ophthalmology. 2013;120:106–114. doi: 10.1016/j.ophtha.2012.07.038. [DOI] [PubMed] [Google Scholar]
  • 45.Criswick V, Schepens C. Familial exudative vitreoretinopathy. Am J Ophthalmol. 1969;68:578–594. doi: 10.1016/0002-9394(69)91237-9. [DOI] [PubMed] [Google Scholar]
  • 46.Chen Z, Battinelli E, Fielder A, et al. A mutation in the norrie disease gene (ndp) associated with x–linked familial exudative vitreoretinopathy. Nat Genet. 1993;5:180–183. doi: 10.1038/ng1093-180. [DOI] [PubMed] [Google Scholar]
  • 47.Shastry B, Hiraoka M, Trese D, Trese M. Norrie disease and exudative vitreoretinopathy in families with affected female carriers. Eur J Ophthalmol. 1999;9:238–242. doi: 10.1177/112067219900900312. [DOI] [PubMed] [Google Scholar]
  • 48.Robitaille J, MacDonald ML, Kaykas A, et al. Mutant frizzled-4 disrupts retinal angiogenesis in familial exudative vitreoretinopathy. Nat Genet. 2002;32:326–330. doi: 10.1038/ng957. [DOI] [PubMed] [Google Scholar]
  • 49.Ye X, Wang Y, Cahill H, et al. Norrin, frizzled-4, and Lrp5 signaling in endothelial cells controls a genetic program for retinal vascularization. Cell. 2009;139:285–298. doi: 10.1016/j.cell.2009.07.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Xia C-H, Liu H, Cheung D, et al. A model for familial exudative vitreoretinopathy caused by LPR5 mutations. Hum Mol Genet. 2008;17:1605–1612. doi: 10.1093/hmg/ddn047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Poulter JA, Ali M, Gilmour DF, et al. Mutations in TSPAN12 cause autosomal-dominant familial exudative vitreoretinopathy. The American Journal of Human Genetics. 2010;86:248–253. doi: 10.1016/j.ajhg.2010.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kwan H, Pecenka V, Tsukamoto A, et al. Transgenes expressing the Wnt-1 and int-2 proto-oncogenes cooperate during mammary carcinogenesis in doubly transgenic mice. Mol Cell Biol. 1992;12:147–154. doi: 10.1128/mcb.12.1.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Jin T. The WNT signalling pathway and diabetes mellitus. Diabetologia. 2008;51:1771–1780. doi: 10.1007/s00125-008-1084-y. [DOI] [PubMed] [Google Scholar]
  • 54.MacDonald M, Goldberg Y, Macfarlane J, Samuels M, Trese M, Shastry B. Genetic variants of frizzled 4 gene in familial exudative vitreoretinopathy and advanced retinopathy of prematurity. Clin Genet. 2005;67:363–366. doi: 10.1111/j.1399-0004.2005.00408.x. [DOI] [PubMed] [Google Scholar]
  • 55.Ells A, Guernsey DL, Wallace K, et al. Severe retinopathy of prematurity associated with FZD4 mutations. Ophthalmic Genet. 2010;31:37–43. doi: 10.3109/13816810903479834. [DOI] [PubMed] [Google Scholar]
  • 56.Kondo H, Kusaka S, Yoshinaga A, Uchio E, Tawara A, Tahira T. Genetic variants of FZD4 and LRP5 genes in patients with advanced retinopathy of prematurity. 2013 [PMC free article] [PubMed] [Google Scholar]
  • 57.Dailey WA, Gryc W, Garg PG, Drenser KA. Frizzled-4 variations associated with retinopathy and intrauterine growth retardation: a potential marker for prematurity and retinopathy. Ophthalmology. 2015;122:1917–1923. doi: 10.1016/j.ophtha.2015.05.036. [DOI] [PubMed] [Google Scholar]
  • 58.Shastry BS, Pendergast SD, Hartzer MK, Liu X, Trese MT. Identification of missense mutations in the Norrie disease gene associated with advanced retinopathy of prematurity. Arch Ophthalmol. 1997;115:651–655. doi: 10.1001/archopht.1997.01100150653015. [DOI] [PubMed] [Google Scholar]
  • 59.Hiraoka M, Berinstein DM, Trese MT, Shastry BS. Insertion and deletion mutations in the dinucleotide repeat region of the Norrie disease gene in patients with advanced retinopathy of prematurity. J Hum Genet. 2001;46:178–181. doi: 10.1007/s100380170085. [DOI] [PubMed] [Google Scholar]
  • 60.Haider M, Devarajan L, Al-Essa M, Kumar H. A C597→ A polymorphism in the Norrie disease gene is associated with advanced retinopathy of prematurity in premature Kuwaiti infants. J Biomed Sci. 2002;9:365–370. doi: 10.1007/BF02256593. [DOI] [PubMed] [Google Scholar]
  • 61.Talks SJ, Ebenezer N, Hykin P, et al. De novo mutations in the 5e 5n the Norrie disease ge Norrie disease gene in retinopathy of prematurity. J Med Genet. 2001;38:e46–e46. doi: 10.1136/jmg.38.12.e46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.John VJ, McClintic JI, Hess DJ, Berrocal AM. Retinopathy of prematurity versus familial exudative vitreoretinopathy: report on clinical and angiographic findings. Ophthalmic Surgery, Lasers and Imaging Retina. 2016;47:14–19. doi: 10.3928/23258160-20151214-02. [DOI] [PubMed] [Google Scholar]
  • 63.Hellstrom A, Perruzzi C, Ju M, et al. Low IGF-I suppresses VEGF-survival signaling in retinal endothelial cells: direct correlation with clinical retinopathy of prematurity. Proceedings of the National Academy of Sciences. 2001;98:5804–5808. doi: 10.1073/pnas.101113998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Powell-Braxton L, Hollingshead P, Warburton C, et al. IGF-I is required for normal embryonic growth in mice. Genes Dev. 1993;7:2609–2617. doi: 10.1101/gad.7.12b.2609. [DOI] [PubMed] [Google Scholar]
  • 65.Ashton I, Zapf J, Einschenk I, MacKenzie I. Insulin-like growth factors (IGF) 1 and 2 in human foetal plasma and relationship to gestational age and foetal size during midpregnancy. Acta Endocrinol (Copenh) 1985;110:558–563. doi: 10.1530/acta.0.1100558. [DOI] [PubMed] [Google Scholar]
  • 66.Hellström A, Engström E, Hård A-L, et al. Postnatal serum insulin-like growth factor I deficiency is associated with retinopathy of prematurity and other complications of premature birth. Pediatrics. 2003;112:1016–1020. doi: 10.1542/peds.112.5.1016. [DOI] [PubMed] [Google Scholar]
  • 67.Hellstrom A, Ley D, Hallberg B, et al. IGF-1 as a drug for preterm infants: a step-wise clinical development. Curr Pharm Des. 2017 doi: 10.2174/1381612823666171002114545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Shire. IGF-1/IGFBP3 Prevention of Retinopathy of Prematurity. Available from: https://clinicaltrials.gov/ct2/show/NCT01096784. NLM identifier: NCT01096784. Accessed 10.10.2017.
  • 69.Bonafè M, Barbieri M, Marchegiani F, et al. Polymorphic variants of insulin-like growth factor I (IGF-I) receptor and phosphoinositide 3-kinase genes affect IGF-I plasma levels and human longevity: cues for an evolutionarily conserved mechanism of life span control. The Journal of Clinical Endocrinology & Metabolism. 2003;88:3299–3304. doi: 10.1210/jc.2002-021810. [DOI] [PubMed] [Google Scholar]
  • 70.Balogh Á, Derzbach L, Vannay Á, Vásárhelyi B. Lack of association between insulin-like growth factor I receptor G+ 3174A polymorphism and retinopathy of prematurity. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2006;244:1035–1038. doi: 10.1007/s00417-005-0203-4. [DOI] [PubMed] [Google Scholar]
  • 71.Shastry B. Assessment of the contribution of insulin-like growth factor I receptor 3174 G–> A polymorphism to the progression of advanced retinopathy of prematurity. Eur J Ophthalmol. 2007;17:950–953. doi: 10.1177/112067210701700613. [DOI] [PubMed] [Google Scholar]
  • 72.Schmetterer L, Polak K. Role of nitric oxide in the control of ocular blood flow. Prog Retin Eye Res. 2001;20:823–847. doi: 10.1016/s1350-9462(01)00014-3. [DOI] [PubMed] [Google Scholar]
  • 73.Ando A, Yang A, Mori K, et al. Nitric oxide is proangiogenic in the retina and choroid. J Cell Physiol. 2002;191:116–124. doi: 10.1002/jcp.10083. [DOI] [PubMed] [Google Scholar]
  • 74.Brooks SE, Gu X, Samuel S, et al. Reduced severity of oxygen-induced retinopathy in eNOS-deficient mice. Invest Ophthalmol Vis Sci. 2001;42:222–228. [PubMed] [Google Scholar]
  • 75.Tsukada T, Yokoyama K, Arai T, et al. Evidence of association of the ecNOS gene polymorphism with plasma NO metabolite levels in humans. Biochem Biophys Res Commun. 1998;245:190–193. doi: 10.1006/bbrc.1998.8267. [DOI] [PubMed] [Google Scholar]
  • 76.Dammann O, Brinkhaus M-J, Bartels DB, et al. Immaturity, perinatal inflammation, and retinopathy of prematurity: a multi-hit hypothesis. Early Hum Dev. 2009;85:325–329. doi: 10.1016/j.earlhumdev.2008.12.010. [DOI] [PubMed] [Google Scholar]
  • 77.Silveira RC, Fortes Filho JB, Procianoy RS. Assessment of the contribution of cytokine plasma levels to detect retinopathy of prematurity in very low birth weight infants. Invest Ophthalmol Vis Sci. 2011;52:1297–1301. doi: 10.1167/iovs.10-6279. [DOI] [PubMed] [Google Scholar]
  • 78.Sato T, Kusaka S, Shimojo H, Fujikado T. Vitreous levels of erythropoietin and vascular endothelial growth factor in eyes with retinopathy of prematurity. Ophthalmology. 2009;116:1599–1603. doi: 10.1016/j.ophtha.2008.12.023. [DOI] [PubMed] [Google Scholar]
  • 79.Matsuda K, Okamoto N, Kondo M, et al. Mast cell hyperactivity underpins the development of oxygen-induced retinopathy. The Journal of clinical investigation. 2017;127:3987–4000. doi: 10.1172/JCI89893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Barde Y-A, Edgar D, Thoenen H. Purification of a new neurotrophic factor from mammalian brain. The EMBO journal. 1982;1:549. doi: 10.1002/j.1460-2075.1982.tb01207.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Hyman C, Hofer M, Barde Y-A, et al. BDNF is a neurotrophic factor for dopaminergic neurons of the substantia nigra. Nature. 1991;350:230–232. doi: 10.1038/350230a0. [DOI] [PubMed] [Google Scholar]
  • 82.Chen H, Weber AJ. BDNF enhances retinal ganglion cell survival in cats with optic nerve damage. Invest Ophthalmol Vis Sci. 2001;42:966–974. [PubMed] [Google Scholar]
  • 83.Loeliger MM, Briscoe T, Rees SM. BDNF increases survival of retinal dopaminergic neurons after prenatal compromise. Invest Ophthalmol Vis Sci. 2008;49:1282–1289. doi: 10.1167/iovs.07-0521. [DOI] [PubMed] [Google Scholar]
  • 84.Sood BG, Madan A, Saha S, et al. Perinatal systemic inflammatory response syndrome and retinopathy of prematurity. Pediatr Res. 2010;67:394. doi: 10.1203/PDR.0b013e3181d01a36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Rao R, Mashburn CB, Mao J, Wadhwa N, Smith GM, Desai NS. Brain-derived neurotrophic factor (BDNF) in infants< 32 weeks gestational age: Correlation with antenatal factors and postnatal outcomes. Pediatr Res. 2009;65:548. doi: 10.1203/PDR.0b013e31819d9ea5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Hellgren G, Willett K, Engstrom E, et al. Proliferative retinopathy is associated with impaired increase in BDNF and RANTES expression levels after preterm birth. Neonatology. 2010;98:409–418. doi: 10.1159/000317779. [DOI] [PubMed] [Google Scholar]
  • 87.Hartnett ME, Morrison MA, Smith S, et al. Genetic Variants Associated With Severe Retinopathy of Prematurity in Extremely Low Birth Weight InfantsGene Variants in ROP in Extremely Low Birth Weight Infants. Invest Ophthalmol Vis Sci. 2014;55:6194–6203. doi: 10.1167/iovs.14-14841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Moravski CJ, Kelly DJ, Cooper ME, et al. Retinal neovascularization is prevented by blockade of the renin-angiotensin system. Hypertension. 2000;36:1099–1104. doi: 10.1161/01.hyp.36.6.1099. [DOI] [PubMed] [Google Scholar]
  • 89.Liang S, Pan M, Hu N, et al. Association of angiotensin-converting enzyme gene 2350 G/A polymorphism with diabetic retinopathy in Chinese Han population. Mol Biol Rep. 2013;40:463–468. doi: 10.1007/s11033-012-2081-2. [DOI] [PubMed] [Google Scholar]
  • 90.Haider M, Devarajan L, Al-Essa M, Kumar H. Angiotensin-converting enzyme gene insertion/deletion polymorphism in Kuwaiti children with retinopathy of prematurity. Neonatology. 2002;82:84–88. doi: 10.1159/000063092. [DOI] [PubMed] [Google Scholar]
  • 91.Mohamed S, Schaa K, Cooper ME, et al. Genetic contributions to the development of retinopathy of prematurity. Pediatr Res. 2009;65:193–197. doi: 10.1203/PDR.0b013e31818d1dbd. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Holash J, Wiegand S, Yancopoulos G. New model of tumor angiogenesis: dynamic balance between vessel regression and growth mediated by angiopoietins and VEGF. Oncogene. 1999;18 doi: 10.1038/sj.onc.1203035. [DOI] [PubMed] [Google Scholar]
  • 93.Lee J, Kim KE, Choi D-K, et al. Angiopoietin-1 Guides Directional Angiogenesis Through Integrin α v β 5 Signaling for Recovery of Ischemic Retinopathy. Sci Transl Med. 2013;5:203ra127–203ra127. doi: 10.1126/scitranslmed.3006666. [DOI] [PubMed] [Google Scholar]
  • 94.Campochiaro PA. Molecular pathogenesis of retinal and choroidal vascular diseases. Prog Retin Eye Res. 2015;49:67–81. doi: 10.1016/j.preteyeres.2015.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Feng Y, Hagen Fv, Pfister F, et al. Impaired pericyte recruitment and abnormal retinal angiogenesis as a result of angiopoietin-2 overexpression. Thromb Haemost. 2007;97:99. [PubMed] [Google Scholar]
  • 96.Sato T, Shima C, Kusaka S. Vitreous levels of angiopoietin-1 and angiopoietin-2 in eyes with retinopathy of prematurity. Am J Ophthalmol. 2011;151:353–357.e351. doi: 10.1016/j.ajo.2010.08.037. [DOI] [PubMed] [Google Scholar]
  • 97.Watanabe D, Suzuma K, Matsui S, et al. Erythropoietin as a retinal angiogenic factor in proliferative diabetic retinopathy. N Engl J Med. 2005;353:782–792. doi: 10.1056/NEJMoa041773. [DOI] [PubMed] [Google Scholar]
  • 98.Chen J, Connor KM, Aderman CM, Smith LE. Erythropoietin deficiency decreases vascular stability in mice. The Journal of clinical investigation. 2008;118:526–533. doi: 10.1172/JCI33813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Yang Z, Wang H, Jiang Y, Hartnett ME. VEGFA activates erythropoietin receptor and enhances VEGFR2-mediated pathological angiogenesis. The American journal of pathology. 2014;184:1230–1239. doi: 10.1016/j.ajpath.2013.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Yang N, Zhang W, He T, Xing Y. Exogenous erythropoietin aggravates retinal neovascularizationin a murine model of proliferative retinopathy. Turkish journal of medical sciences. 2017;47:1642–1650. doi: 10.3906/sag-1609-49. [DOI] [PubMed] [Google Scholar]
  • 101.Semenza GL. Oxygen sensing, homeostasis, and disease. N Engl J Med. 2011;365:537–547. doi: 10.1056/NEJMra1011165. [DOI] [PubMed] [Google Scholar]
  • 102.Lin M, Chen Y, Jin J, et al. Ischaemia-induced retinal neovascularisation and diabetic retinopathy in mice with conditional knockout of hypoxia-inducible factor-1 in retinal Müller cells. Diabetologia. 2011;54:1554–1566. doi: 10.1007/s00125-011-2081-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hoppe G, Yoon S, Gopalan B, et al. Comparative systems pharmacology of HIF stabilization in the prevention of retinopathy of prematurity. Proceedings of the National Academy of Sciences. 2016;113:E2516–E2525. doi: 10.1073/pnas.1523005113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Morita M, Ohneda O, Yamashita T, et al. HLF/HIF 2α is a key factor in retinopathy of prematurity in association with erythropoietin. The EMBO Journal. 2003;22:1134–1146. doi: 10.1093/emboj/cdg117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Huang B, Deora AB, He K-L, et al. Hypoxia-inducible factor-1 drives annexin A2 system-mediated perivascular fibrin clearance in oxygen-induced retinopathy in mice. Blood. 2011;118:2918–2929. doi: 10.1182/blood-2011-03-341214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Rankin EB, Biju MP, Liu Q, et al. Hypoxia-inducible factor–2 (HIF-2) regulates hepatic erythropoietin in vivo. J Clin Invest. 2007;117:1068. doi: 10.1172/JCI30117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wolfsberg TG, Primakoff P, Myles DG, White J. ADAM, a novel family of membrane proteins containing A Disintegrin And Metalloprotease domain: multipotential functions in cell-cell and cell-matrix interactions. The Journal of cell biology. 1995;131:275–278. doi: 10.1083/jcb.131.2.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Weskamp G, Mendelson K, Swendeman S, et al. Pathological neovascularization is reduced by inactivation of ADAM17 in endothelial cells but not in pericytes. Circ Res. 2010;106:932–940. doi: 10.1161/CIRCRESAHA.109.207415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Guaiquil VH, Hewing NJ, Chiang MF, Rosenblatt MI, Chan RP, Blobel CP. A Murine Model for Retinopathy of Prematurity Identifies Endothelial Cell Proliferation as a Potential Mechanism for Plus DiseaseIncreased Proliferation of Vascular Cells in Plus Disease. Invest Ophthalmol Vis Sci. 2013;54:5294–5302. doi: 10.1167/iovs.12-11492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Mahmoodi M, Sahebjam S, Smookler D, Khokha R, Mort JS. Lack of tissue inhibitor of metalloproteinases-3 results in an enhanced inflammatory response in antigen-induced arthritis. The American journal of pathology. 2005;166:1733–1740. doi: 10.1016/S0002-9440(10)62483-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Manolio TA, Collins FS, Cox NJ, et al. Finding the missing heritability of complex diseases. Nature. 2009;461:747–753. doi: 10.1038/nature08494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Darlow BA, Lui K, Kusuda S, et al. International variations and trends in the treatment for retinopathy of prematurity. Br J Ophthalmol. 2017 doi: 10.1136/bjophthalmol-2016-310041. bjophthalmol-2016-310041. [DOI] [PubMed] [Google Scholar]
  • 113.Mora JS, Waite C, Gilbert CE, Breidenstein B, Sloper JJ. A worldwide survey of retinopathy of prematurity screening. Br J Ophthalmol. 2017 doi: 10.1136/bjophthalmol-2017-310709. bjophthalmol-2017-310709. [DOI] [PubMed] [Google Scholar]
  • 114.Moleta C, Campbell JP, Kalpathy-Cramer J, et al. Plus Disease in Retinopathy of Prematurity: Diagnostic Trends in 2016 Versus 2007. Am J Ophthalmol. 2017;176:70–76. doi: 10.1016/j.ajo.2016.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Manja V, Lakshminrusimha S, Cook DJ. Oxygen saturation target range for extremely preterm infants: a systematic review and meta-analysis. JAMA pediatrics. 2015;169:332–340. doi: 10.1001/jamapediatrics.2014.3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Meng Q, Huang L, Sun Y, et al. Effect of high-density lipoprotein metabolic pathway gene variations and risk factors on neovascular age-related macular degeneration and polypoidal choroidal vasculopathy in China. PLoS One. 2015;10:e0143924. doi: 10.1371/journal.pone.0143924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Helgason H, Sulem P, Duvvari MR, et al. A rare nonsynonymous sequence variant in C3 is associated with high risk of age-related macular degeneration. Nat Genet. 2013;45:1371–1374. doi: 10.1038/ng.2740. [DOI] [PubMed] [Google Scholar]
  • 118.AMD Gene Consortium. Seven new loci associated with age-related macular degeneration. Nat Genet. 2013;45:433–439. doi: 10.1038/ng.2578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Burdon KP, Fogarty RD, Shen W, et al. Genome-wide association study for sight-threatening diabetic retinopathy reveals association with genetic variation near the GRB2 gene. Diabetologia. 2015;58:2288–2297. doi: 10.1007/s00125-015-3697-2. [DOI] [PubMed] [Google Scholar]
  • 120.Sheu WH-H, Kuo JZ, Lee I-T, et al. Genome-wide association study in a Chinese population with diabetic retinopathy. Hum Mol Genet. 2013;22:3165–3173. doi: 10.1093/hmg/ddt161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Khor CC, Do T, Jia H, et al. Genome-wide association study identifies five new susceptibility loci for primary angle closure glaucoma. Nat Genet. 2016;48:556–562. doi: 10.1038/ng.3540. [DOI] [PubMed] [Google Scholar]
  • 122.Bailey JNC, Loomis SJ, Kang JH, et al. Genome-wide association analysis identifies TXNRD2, ATXN2 and FOXC1 as susceptibility loci for primary open angle glaucoma. Nat Genet. 2016;48:189. doi: 10.1038/ng.3482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Vithana EN, Khor C-C, Qiao C, et al. Genome-wide association analyses identify three new susceptibility loci for primary angle closure glaucoma. Nat Genet. 2012;44:1142–1146. doi: 10.1038/ng.2390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Verhoeven VJ, Hysi PG, Wojciechowski R, et al. Genome-wide meta-analyses of multiancestry cohorts identify multiple new susceptibility loci for refractive error and myopia. Nat Genet. 2013;45:314–318. doi: 10.1038/ng.2554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Solouki AM, Verhoeven VJ, Van Duijn CM, et al. A genome-wide association study identifies a susceptibility locus for refractive errors and myopia at 15q14. Nat Genet. 2010;42:897. doi: 10.1038/ng.663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hysi PG, Young TL, Mackey DA, et al. A genome-wide association study for myopia and refractive error identifies a susceptibility locus at 15q25. Nat Genet. 2010;42:902–905. doi: 10.1038/ng.664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Syreeni A, El-Osta A, Forsblom C, et al. Genetic examination of SETD7 and SUV39H1/H2 methyltransferases and the risk of diabetes complications in patients with type 1 diabetes. Diabetes. 2011;60:3073–3080. doi: 10.2337/db11-0073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Zhong Q, Kowluru RA. Epigenetic changes in mitochondrial superoxide dismutase in the retina and the development of diabetic retinopathy. Diabetes. 2011;60:1304–1313. doi: 10.2337/db10-0133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.McArthur K, Feng B, Wu Y, Chen S, Chakrabarti S. MicroRNA-200b regulates vascular endothelial growth factor–mediated alterations in diabetic retinopathy. Diabetes. 2011;60:1314–1323. doi: 10.2337/db10-1557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Kowluru RA, Santos JM, Mishra M. Epigenetic modifications and diabetic retinopathy. BioMed research international. 2013;2013 doi: 10.1155/2013/635284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Zhong Q, Kowluru RA. Regulation of matrix metalloproteinase-9 by epigenetic modifications and the development of diabetic retinopathy. Diabetes. 2013;62:2559–2568. doi: 10.2337/db12-1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Psaty BM, O’donnell CJ, Gudnason V, et al. Cohorts for heart and aging research in genomic epidemiology (CHARGE) consortium. Circ Cardiovasc Genet. 2009;2:73–80. doi: 10.1161/CIRCGENETICS.108.829747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Lu Y, Vitart V, Burdon KP, et al. Genome-wide association analyses identify multiple loci associated with central corneal thickness and keratoconus. Nat Genet. 2013;45:155–163. doi: 10.1038/ng.2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Jensen RA, Sim X, Smith AV, et al. Novel genetic loci associated with retinal microvascular diameter. Circ Cardiovasc Genet. 2015 doi: 10.1161/CIRCGENETICS.115.001142. CIRCGENETICS. 115.001142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.National Institute of Environmental Health Sciences. Genes Associated With Bronchopulmonary Dysplasia and Retinopathy of Prematurity. Available from: https://clinicaltrials.gov/ct2/show/NCT01780155. NLM identifier: NCT01780155. Accessed 05.10.17.
  • 136.Imaging and Informatics in ROP Research Consortium. Imaging and Informatics in Retinopathy of Prematurity. 2017 http://i-rop.github.io/. 2017 Accessed 10.10.17.
  • 137.Vannay Á, Dunai G, Bányász I, et al. Association of genetic polymorphisms of vascular endothelial growth factor and risk for proliferative retinopathy of prematurity. Pediatr Res. 2005;57:396–398. doi: 10.1203/01.PDR.0000153867.80238.E0. [DOI] [PubMed] [Google Scholar]
  • 138.Kaya M, Çokakli M, Berk AT, et al. Associations of VEGF/VEGF-Receptor and HGF/c-Met promoter polymorphisms with progression/regression of retinopathy of prematurity. Curr Eye Res. 2013;38:137–142. doi: 10.3109/02713683.2012.731550. [DOI] [PubMed] [Google Scholar]
  • 139.Shastry BS, Qu X. Lack of association of the VEGF gene promoter (−634 G→ C and−460 C→ T) polymorphism and the risk of advanced retinopathy of prematurity. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2007;245:741–743. doi: 10.1007/s00417-006-0480-6. [DOI] [PubMed] [Google Scholar]
  • 140.Kusuda T, Hikino S, Ohga S, et al. Genetic variation of vascular endothelial growth factor pathway does not correlate with the severity of retinopathy of prematurity. J Perinatol. 2011;31:246–250. doi: 10.1038/jp.2010.111. [DOI] [PubMed] [Google Scholar]
  • 141.Kwinta P, Bik-Multanowski M, Mitkowska Z, Tomasik T, Pietrzyk JJ. The clinical role of vascular endothelial growth factor (VEGF) system in the pathogenesis of retinopathy of prematurity. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2008;246:1467–1475. doi: 10.1007/s00417-008-0865-9. [DOI] [PubMed] [Google Scholar]
  • 142.Kalmeh ZA, Azarpira N, Mosallaei M, Hosseini H, Malekpour Z. Genetic polymorphisms of vascular endothelial growth factor and risk for retinopathy of prematurity in South of Iran. Mol Biol Rep. 2013;40:4613–4618. doi: 10.1007/s11033-013-2554-y. [DOI] [PubMed] [Google Scholar]
  • 143.Gismondi D, Ndoja L, Qu X, Shastry BS. Lack of association of VEGF gene 3′-UTR polymorphisms (C702T, C936T and G1612A) and the risk of developing advanced retinopathy of prematurity (ROP) Graefe’s Archive for Clinical and Experimental Ophthalmology. 2012:1–3. doi: 10.1007/s00417-011-1913-4. [DOI] [PubMed] [Google Scholar]
  • 144.Shastry BS. Lack of association of VEGF (−2578 C→ A) and ANG 2 (−35 G→ C) gene polymorphisms with the progression of retinopathy of prematurity. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2009;247:859–860. doi: 10.1007/s00417-008-0988-z. [DOI] [PubMed] [Google Scholar]
  • 145.Bányász I, Bokodi G, Vannay Á, et al. Genetic polymorphisms of vascular endothelial growth factor and angiopoietin 2 in retinopathy of prematurity. Curr Eye Res. 2006;31:685–690. doi: 10.1080/02713680600801123. [DOI] [PubMed] [Google Scholar]
  • 146.Hartnett ME, Cotten CM. Genomics in the neonatal nursery: Focus on ROP. Paper presented at: Semin Perinatol. 2015 doi: 10.1053/j.semperi.2015.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Hiraoka M, Takahashi H, Orimo H, Hiraoka M, Ogata T, Azuma N. Genetic screening of Wnt signaling factors in advanced retinopathy of prematurity. Mol Vis. 2010;16:2572–7. [PMC free article] [PubMed] [Google Scholar]
  • 148.Rusai K, Vannay A, Szebeni B, et al. Endothelial nitric oxide synthase gene T-786C and 27-bp repeat gene polymorphisms in retinopathy of prematurity. Mol Vis. 2008;14:286–290. [PMC free article] [PubMed] [Google Scholar]
  • 149.Yanamandra K, Napper D, Pramanik A, Bocchini JA, Jr, Dhanireddy R. Endothelial nitric oxide synthase genotypes in the etiology of retinopathy of prematurity in premature infants. Ophthalmic Genet. 2010;31:173–177. doi: 10.3109/13816810.2010.497528. [DOI] [PubMed] [Google Scholar]
  • 150.Shastry BS. Endothelial nitric oxide synthase gene promoter polymorphism (T-786C) may be associated with advanced retinopathy of prematurity. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2013;251:2251. doi: 10.1007/s00417-012-2231-1. [DOI] [PubMed] [Google Scholar]
  • 151.Hutcheson KA, Paluru PC, Bernstein SL, et al. Norrie disease gene sequence variants in an ethnically diverse population with retinopathy of prematurity. Mol Vis. 2005;11:501–508. [PubMed] [Google Scholar]
  • 152.Dickinson JL, Sale MM, Passmore A, et al. Mutations in the NDP gene: contribution to Norrie disease, familial exudative vitreoretinopathy and retinopathy of prematurity. Clin Experiment Ophthalmol. 2006;34:682–688. doi: 10.1111/j.1442-9071.2006.01314.x. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

supplement

RESOURCES